Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
ACS Chem Neurosci ; 15(14): 2633-2642, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38967483

ABSTRACT

In order to investigate the effectiveness and safety of miR-23b-3p in anti-seizure activity and to elucidate the regulatory relationship between miR-23b-3p and Cx43 in the nervous system, we have established a lithium chloride-pilocarpine (PILO) status epilepticus (SE) model. Rats were randomly divided into the following groups: seizure control (PILO), valproate sodium (VPA+PILO), recombinant miR-23b-3p overexpression (miR+PILO), miR-23b-3p sponges (Sponges+PILO), and scramble sequence negative control (Scramble+PILO) (n = 6/group). After experiments, we got the following results. In the acute phase, the time required for rats to reach stage IV after PILO injection was significantly longer in VPA+PILO and miR+PILO. In the chronic phase after SE, the frequency of spontaneous recurrent seizures (SRSs) in VPA+PILO and miR+PILO was significantly reduced. At 10 min before seizure cessation, the average energy expression of fast ripples (FRs) in VPA+PILO and miR+PILO was significantly lower than in PILO. After 28 days of seizure, Cx43 expression in PILO was significantly increased, and Beclin1expression in all groups was significantly increased. After 28 days of SE,the number of synapses in the CA1 region of the hippocampus was significantly higher in the VPA+PILO and miR+PILO groups compared to that in the PILO group. After 28 days of SE ,hippocampal necrotic cells in the CA3 region were significantly lower in the VPA+PILO and miR+PILO groups compared to those in the PILO group. There were no significant differences in biochemical indicators among the experimental group rats 28 days after SE compared to the seizure control group. Based on the previous facts, we can reach the conclusion that MiR-23b-3p targets and blocks the expression of hippocampal Cx43 which can reduce the formation of pathological FRs, thereby alleviating the severity of seizures, improving seizure-induced brain damage.


Subject(s)
Connexin 43 , Hippocampus , MicroRNAs , Rats, Sprague-Dawley , Status Epilepticus , Animals , Male , Rats , Brain Injuries/metabolism , Connexin 43/metabolism , Connexin 43/genetics , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/drug effects , MicroRNAs/metabolism , MicroRNAs/genetics , Pilocarpine/toxicity , Seizures/metabolism , Seizures/chemically induced , Status Epilepticus/chemically induced , Status Epilepticus/metabolism
2.
Life Sci ; 352: 122917, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39019341

ABSTRACT

BACKGROUND: Status epilepticus (SE) as a severe neurodegenerative disease, greatly negatively affects people's health, and there is an urgent need for innovative treatments. The valuable neuroprotective effects of glucagon-like peptide-1 (GLP-1) in several neurodegenerative diseases have raised motivation to investigate the dipeptidyl peptidase-4 (DPP-4) inhibitor; alogliptin (ALO), an oral antidiabetic drug as a potential treatment for SE. ALO has shown promising neuroprotective effects in Alzheimer's and Parkinson's diseases, but its impact on SE has not yet been studied. AIM: The present study aimed to explore the repurposing potential for ALO in a lithium/pilocarpine (Li/Pil)-induced SE model in rats. MAIN METHODS: ALO (30 mg/kg/day) was administered via gavage for 14 days, and SE was subsequently induced in the rats using a single dose of Li/Pil (127/60 mg/kg), while levetiracetam was used as a standard antiepileptic drug. KEY FINDINGS: The results showed that ALO reduced seizure severity and associated hippocampal neurodegeneration. ALO also increased γ-aminobutyric acid (GABA) levels, diminished glutamate spikes, and corrected glial fibrillary acidic protein (GFAP) changes. At the molecular level, ALO increased GLP-1 levels and activated its downstream signaling pathway, AMP-activated protein kinase (AMPK)/sirtuin-1 (SIRT1). ALO also dampened the brain's pro-oxidant response, curbed neuroinflammation, and counteracted hippocampal apoptosis affording neuroprotection. In addition, it activated autophagy as indicated by Beclin1 elevation. SIGNIFICANCE: This study suggested that the neuroprotective properties and autophagy-enhancing effects of ALO make it a promising treatment for SE and can potentially be used as a management approach for this condition.


Subject(s)
Autophagy , NF-E2-Related Factor 2 , Neuroprotective Agents , Pilocarpine , Piperidines , Seizures , Sirtuin 1 , Animals , Rats , Sirtuin 1/metabolism , Neuroprotective Agents/pharmacology , Male , Autophagy/drug effects , Piperidines/pharmacology , NF-E2-Related Factor 2/metabolism , Seizures/chemically induced , Seizures/drug therapy , Seizures/metabolism , Lithium/pharmacology , AMP-Activated Protein Kinases/metabolism , Uracil/analogs & derivatives , Uracil/pharmacology , Rats, Sprague-Dawley , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Status Epilepticus/metabolism , Signal Transduction/drug effects
3.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38892448

ABSTRACT

Canonical transient receptor potential channel 3 (TRPC3) is the most abundant TRPC channel in the brain and is highly expressed in all subfields of the hippocampus. Previous studies have suggested that TRPC3 channels may be involved in the hyperexcitability of hippocampal pyramidal neurons and seizures. Genetic ablation of TRPC3 channel expression reduced the intensity of pilocarpine-induced status epilepticus (SE). However, the underlying cellular mechanisms remain unexplored and the contribution of TRPC3 channels to SE-induced neurodegeneration is not determined. In this study, we investigated the contribution of TRPC3 channels to the electrophysiological properties of hippocampal pyramidal neurons and hippocampal synaptic plasticity, and the contribution of TRPC3 channels to seizure-induced neuronal cell death. We found that genetic ablation of TRPC3 expression did not alter basic electrophysiological properties of hippocampal pyramidal neurons and had a complex impact on epileptiform bursting in CA3. However, TRPC3 channels contribute significantly to long-term potentiation in CA1 and SE-induced neurodegeneration. Our results provided further support for therapeutic potential of TRPC3 inhibitors and raised new questions that need to be answered by future studies.


Subject(s)
Cell Death , Hippocampus , Pyramidal Cells , Seizures , TRPC Cation Channels , Animals , TRPC Cation Channels/metabolism , TRPC Cation Channels/genetics , Mice , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Hippocampus/metabolism , Hippocampus/pathology , Seizures/metabolism , Seizures/pathology , Status Epilepticus/metabolism , Status Epilepticus/pathology , Status Epilepticus/chemically induced , Male , Neurons/metabolism , Pilocarpine , Long-Term Potentiation , Mice, Knockout , Mice, Inbred C57BL , Neuronal Plasticity
4.
eNeuro ; 11(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38937107

ABSTRACT

γ-Aminobutyric acid (GABA) is the principal inhibitory neurotransmitter in the adult brain which mediates its rapid effects on neuronal excitability via ionotropic GABAA receptors. GABA levels in the brain are critically dependent upon GABA-aminotransferase (GABA-AT) which promotes its degradation. Vigabatrin, a low-affinity GABA-AT inhibitor, exhibits anticonvulsant efficacy, but its use is limited due to cumulative ocular toxicity. OV329 is a rationally designed, next-generation GABA-AT inhibitor with enhanced potency. We demonstrate that sustained exposure to OV329 in mice reduces GABA-AT activity and subsequently elevates GABA levels in the brain. Parallel increases in the efficacy of GABAergic inhibition were evident, together with elevations in electroencephalographic delta power. Consistent with this, OV329 exposure reduced the severity of status epilepticus and the development of benzodiazepine refractory seizures. Thus, OV329 may be of utility in treating seizure disorders and associated pathologies that result from neuronal hyperexcitability.


Subject(s)
4-Aminobutyrate Transaminase , Anticonvulsants , Benzodiazepines , Seizures , gamma-Aminobutyric Acid , Animals , Anticonvulsants/pharmacology , Anticonvulsants/administration & dosage , Male , Benzodiazepines/pharmacology , 4-Aminobutyrate Transaminase/antagonists & inhibitors , 4-Aminobutyrate Transaminase/metabolism , Seizures/drug therapy , Seizures/metabolism , gamma-Aminobutyric Acid/metabolism , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Brain/drug effects , Brain/metabolism , Neural Inhibition/drug effects , Neural Inhibition/physiology , Mice , Electroencephalography , Disease Models, Animal , Status Epilepticus/drug therapy , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Female
5.
ACS Chem Neurosci ; 15(10): 1951-1966, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38696478

ABSTRACT

Aims: the study aimed to (i) use adeno-associated virus technology to modulate parvalbumin (PV) gene expression, both through overexpression and silencing, within the hippocampus of male mice and (ii) assess the impact of PV on the metabolic pathway of glutamate and γ-aminobutyric acid (GABA). Methods: a status epilepticus (SE) mouse model was established by injecting kainic acid into the hippocampus of transgenic mice. When the seizures of mice reached SE, the mice were killed at that time point and 30 min after the onset of SE. Hippocampal tissues were extracted and the mRNA and protein levels of PV and the 65 kDa (GAD65) and 67 kDa (GAD67) isoforms of glutamate decarboxylase were assessed using real-time quantitative polymerase chain reaction and Western blot, respectively. The concentrations of glutamate and GABA were detected with high-performance liquid chromatography (HPLC), and the intracellular calcium concentration was detected using flow cytometry. Results: we demonstrate that the expression of PV is associated with GAD65 and GAD67 and that PV regulates the levels of GAD65 and GAD67. PV was correlated with calcium concentration and GAD expression. Interestingly, PV overexpression resulted in a reduction in calcium ion concentration, upregulation of GAD65 and GAD67, elevation of GABA concentration, reduction in glutamate concentration, and an extension of seizure latency. Conversely, PV silencing induced the opposite effects. Conclusion: parvalbumin may affect the expression of GAD65 and GAD67 by regulating calcium ion concentration, thereby affecting the metabolic pathways associated with glutamate and GABA. In turn, this contributes to the regulation of seizure activity.


Subject(s)
Calcium , Glutamic Acid , Kainic Acid , Parvalbumins , Status Epilepticus , gamma-Aminobutyric Acid , Animals , Male , Mice , Calcium/metabolism , Disease Models, Animal , gamma-Aminobutyric Acid/metabolism , Glutamate Decarboxylase/metabolism , Glutamic Acid/metabolism , Hippocampus/metabolism , Mice, Transgenic , Parvalbumins/metabolism , Status Epilepticus/metabolism , Status Epilepticus/chemically induced
6.
Redox Biol ; 73: 103168, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38714094

ABSTRACT

Glutathione (GSH) is a major endogenous antioxidant, and its depletion has been observed in several brain diseases including epilepsy. Previous studies in our laboratory have shown that dimercaprol (DMP) can elevate GSH via post-translational activation of glutamate cysteine ligase (GCL), the rate limiting GSH biosynthetic enzyme and inhibit neuroinflammation in vitro. Here we determined 1) the role of cysteamine as a new mechanism by which DMP increases GSH biosynthesis and 2) its ability to inhibit neuroinflammation and neuronal injury in the rat kainate model of epilepsy. DMP depleted cysteamine in a time- and concentration-dependent manner in a cell free system. To guide the in vivo administration of DMP, its pharmacokinetic profile was determined in the plasma, liver, and brain. The results confirmed DMP's ability to cross the blood-brain-barrier. Treatment of rats with DMP (30 mg/kg) depleted cysteamine in the liver and hippocampus that was associated with increased GCL activity in these tissues. GSH levels were significantly increased (20 %) in the hippocampus 1 h after 30 mg/kg DMP administration. Following DMP (30 mg/kg) administration once daily, a marked attenuation of GSH depletion was seen in the SE model. SE-induced inflammatory markers including cytokine release, microglial activation, and neuronal death were significantly attenuated in the hippocampus with DMP treatment. Taken together, these results highlight the importance of restoring redox status with rescue of GSH depletion by DMP in post epileptogenic insults.


Subject(s)
Glutathione , Neuroinflammatory Diseases , Oxidative Stress , Status Epilepticus , Animals , Rats , Glutathione/metabolism , Status Epilepticus/metabolism , Status Epilepticus/drug therapy , Oxidative Stress/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/etiology , Male , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/drug effects , Cysteamine/pharmacology , Antioxidants/pharmacology , Antioxidants/metabolism , Glutamate-Cysteine Ligase/metabolism , Liver/metabolism , Liver/pathology , Liver/drug effects
7.
Brain Behav Immun ; 120: 121-140, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38777288

ABSTRACT

BACKGROUND: The purinergic ATP-gated P2X7 receptor (P2X7R) is increasingly recognized to contribute to pathological neuroinflammation and brain hyperexcitability. P2X7R expression has been shown to be increased in the brain, including both microglia and neurons, in experimental models of epilepsy and patients. To date, the cell type-specific downstream effects of P2X7Rs during seizures remain, however, incompletely understood. METHODS: Effects of P2X7R signaling on seizures and epilepsy were analyzed in induced seizure models using male mice including the kainic acid model of status epilepticus and pentylenetetrazole model and in male and female mice in a genetic model of Dravet syndrome. RNA sequencing was used to analyze P2X7R downstream signaling during seizures. To investigate the cell type-specific role of the P2X7R during seizures and epilepsy, we generated mice lacking exon 2 of the P2rx7 gene in either microglia (P2rx7:Cx3cr1-Cre) or neurons (P2rx7:Thy-1-Cre). To investigate the protective potential of overexpressing P2X7R in GABAergic interneurons, P2X7Rs were overexpressed using adeno-associated virus transduction under the mDlx promoter. RESULTS: RNA sequencing of hippocampal tissue from wild-type and P2X7R knock-out mice identified both glial and neuronal genes, in particular genes involved in GABAergic signaling, under the control of the P2X7R following seizures. Mice with deleted P2rx7 in microglia displayed less severe acute seizures and developed a milder form of epilepsy, and microglia displayed an anti-inflammatory molecular profile. In contrast, mice lacking P2rx7 in neurons showed a more severe seizure phenotype when compared to epileptic wild-type mice. Analysis of single-cell expression data revealed that human P2RX7 expression is elevated in the hippocampus of patients with temporal lobe epilepsy in excitatory and inhibitory neurons. Functional studies determined that GABAergic interneurons display increased responses to P2X7R activation in experimental epilepsy. Finally, we show that viral transduction of P2X7R in GABAergic interneurons protects against evoked and spontaneous seizures in experimental temporal lobe epilepsy and in mice lacking Scn1a, a model of Dravet syndrome. CONCLUSIONS: Our results suggest a dual and opposing action of P2X7R in epilepsy and suggest P2X7R overexpression in GABAergic interneurons as a novel therapeutic strategy for acquired and, possibly, genetic forms of epilepsy.


Subject(s)
Disease Models, Animal , Microglia , Neurons , Receptors, Purinergic P2X7 , Seizures , Animals , Microglia/metabolism , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2X7/genetics , Male , Mice , Seizures/metabolism , Seizures/genetics , Neurons/metabolism , Female , Mice, Inbred C57BL , Kainic Acid , Epilepsies, Myoclonic/metabolism , Epilepsies, Myoclonic/genetics , Hippocampus/metabolism , Status Epilepticus/metabolism , Status Epilepticus/genetics , Mice, Knockout , Pentylenetetrazole , Signal Transduction , GABAergic Neurons/metabolism , Epilepsy/metabolism , Epilepsy/genetics , Brain/metabolism
8.
eNeuro ; 11(5)2024 May.
Article in English | MEDLINE | ID: mdl-38641413

ABSTRACT

The mechanisms by which brain insults lead to subsequent epilepsy remain unclear. Insults including trauma, stroke, infections, and long seizures (status epilepticus, SE) increase the nuclear expression and chromatin binding of the neuron-restrictive silencing factor/RE-1 silencing transcription factor (NRSF/REST). REST/NRSF orchestrates major disruption of the expression of key neuronal genes, including ion channels and neurotransmitter receptors, potentially contributing to epileptogenesis. Accordingly, transient interference with REST/NRSF chromatin binding after an epilepsy-provoking SE suppressed spontaneous seizures for the 12 d duration of a prior study. However, whether the onset of epileptogenesis was suppressed or only delayed has remained unresolved. The current experiments determined if transient interference with REST/NRSF chromatin binding prevented epileptogenesis enduringly or, alternatively, slowed epilepsy onset. Epileptogenesis was elicited in adult male rats via systemic kainic acid-induced SE (KA-SE). We then determined if decoy, NRSF-binding-motif oligodeoxynucleotides (NRSE-ODNs), given twice following KA-SE (1) prevented REST/NRSF binding to chromatin, using chromatin immunoprecipitation, or (2) prevented the onset of spontaneous seizures, measured with chronic digital video-electroencephalogram. Blocking NRSF function transiently after KA-SE significantly lengthened the latent period to a first spontaneous seizure. Whereas this intervention did not influence the duration and severity of spontaneous seizures, total seizure number and seizure burden were lower in the NRSE-ODN compared with scrambled-ODN cohorts. Transient interference with REST/NRSF function after KA-SE delays and moderately attenuates insult-related hippocampal epilepsy, but does not abolish it. Thus, the anticonvulsant and antiepileptogenic actions of NRSF are but one of the multifactorial mechanisms generating epilepsy in the adult brain.


Subject(s)
Chromatin , Kainic Acid , Rats, Sprague-Dawley , Animals , Male , Chromatin/metabolism , Kainic Acid/pharmacology , Repressor Proteins/metabolism , Repressor Proteins/genetics , Status Epilepticus/metabolism , Disease Models, Animal , Hippocampus/metabolism , Rats , Epilepsy/metabolism
9.
J Int Med Res ; 52(3): 3000605241235589, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38546233

ABSTRACT

OBJECTIVE: To investigate the effects of hydrogen therapy on epileptic seizures in rats with refractory status epilepticus and the underlying mechanisms. METHODS: Status epilepticus was induced using pilocarpine. The effects of hydrogen treatment on epilepsy severity in model rats were then monitored using Racine scores and electroencephalography (EEG), followed by western blot of plasma membrane N-methyl-D-aspartate receptor subtype 2B (NR2B) and phosphorylated NR2B expression. We also generated a cellular epilepsy model using Mg2+-free medium and used polymerase chain reaction to investigate the neuroprotective effects of hydrogen. RESULTS: There were no significant differences in Racine scores between the hydrogen and control groups. EEG amplitudes were lower in the hydrogen treatment group than in the control group. In epilepsy model rats, hippocampal cell membrane NR2B expression and phosphorylation increased gradually over time. Although hippocampal cell membrane NR2B expression was not significantly different between the two groups, NR2B phosphorylation levels were significantly lower in the hydrogen group. Hydrogen treatment also increased superoxide dismutase, mitochondrial (SOD2) expression. CONCLUSIONS: Hydrogen treatment reduced EEG amplitudes and NR2B phosphorylation; it also decreased neuronal death by reducing oxidative stress. Hydrogen may thus be a potential treatment for refractory status epilepticus by inhibiting membrane NR2B phosphorylation and oxidative stress.


Subject(s)
Epilepsy , Status Epilepticus , Rats , Animals , Rats, Sprague-Dawley , Phosphorylation , Status Epilepticus/drug therapy , Status Epilepticus/metabolism , Electroencephalography , Oxidative Stress , Hippocampus , Disease Models, Animal
10.
Neurochem Int ; 175: 105717, 2024 May.
Article in English | MEDLINE | ID: mdl-38447759

ABSTRACT

OBJECTIVES: Monoacylglycerol lipase (MAGL) is a cytosolic serine hydrolase considered a potential novel drug target for the treatment of CNS disorders including epilepsy. Here we examined MAGL levels in a rat model of epilepsy. METHODS: Autoradiography has been used to validate the binding properties of the MAGL radiotracer, [3H]T-401, in the rat brain, and to explore spatial and temporal changes in binding levels in a model of temporal lobe epilepsy model using unilateral intra-hippocampal injections of kainic acid (KA) in rats. RESULTS: Specific and saturable binding of [3H]T-401 was detected in both cortical grey and subcortical white matter. Saturation experiments revealed a KD in the range between 15 nM and 17 nM, and full saturation was achieved at concentrations around 30 nM. The binding could be completely blocked with the cold ligand (Ki 44.2 nM) and at higher affinity (Ki 1.27 nM) with another structurally different MAGL inhibitor, ABD 1970. Bilateral reduction in [3H]T-401 binding was observed in the cerebral cortex and the hippocampus few days after status epilepticus that further declined to a level of around 30% compared to the control. No change in binding was observed in either the hypothalamus nor the white matter at any time point. Direct comparison to [3H]UCB-J binding to synaptic vesicle glycoprotein 2 A (SV2A), another protein localized in the pre-synapse, revealed that while binding to MAGL remained low in the chronic phase, SV2A was increased significantly in some cortical areas. SIGNIFICANCE: These data show that MAGL is reduced in the cerebral cortex and hippocampus in a chronic epilepsy model and indicate that MAGL inhibitors may further reduce MAGL activity in the treatment resistant epilepsy patient.


Subject(s)
Epilepsy , Status Epilepticus , Humans , Rats , Animals , Brain/diagnostic imaging , Brain/metabolism , Monoacylglycerol Lipases , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Epilepsy/metabolism , Enzyme Inhibitors/pharmacology
11.
J Cell Physiol ; 239(5): e31249, 2024 May.
Article in English | MEDLINE | ID: mdl-38501376

ABSTRACT

The hippocampal dentate gyrus, responds to diverse pathological stimuli through neurogenesis. This phenomenon, observed following brain injury or neurodegeneration, is postulated to contribute to neuronal repair and functional recovery, thereby presenting an avenue for endogenous neuronal restoration. This study investigated the extent of regenerative response in hippocampal neurogenesis by leveraging the well-established kainic acid-induced status epilepticus model in vivo. In our study, we observed the activation and proliferation of neuronal progenitors or neural stem cell (NSC) and their subsequent migration to the injury sites following the seizure. At the injury sites, new neurons (Tuj1+BrdU+ and NeuN+BrdU+) have been generated indicating regenerative and reparative roles of the progenitor cells. We further detected whether this transient neurogenic burst, which might be a response towards an attempt to repair the brain, is associated with persistent long-term exhaustion of the dentate progenitor cells and impairment of adult neurogenesis marked by downregulation of Ki67, HoPX, and Sox2 with BrdU+ cell in the later part of life. Our studies suggest that the adult brain has the constitutive endogenous regenerative potential for brain repair to restore the damaged neurons, meanwhile, in the long term, it accelerates the depletion of the finite NSC pool in the hippocampal neurogenic niche by changing its proliferative and neurogenic capacity. A thorough understanding of the impact of modulating adult neurogenesis will eventually be required to design novel therapeutics to stimulate or assist brain repair while simultaneously preventing the adverse effects of early robust neurogenesis on the proliferative potential of endogenous neuronal progenitors.


Subject(s)
Hippocampus , Neural Stem Cells , Neurogenesis , Animals , Neural Stem Cells/metabolism , Hippocampus/pathology , Hippocampus/metabolism , Cell Proliferation , Male , Stem Cell Niche , Dentate Gyrus/pathology , Dentate Gyrus/physiopathology , Neurons/metabolism , Neurons/pathology , Kainic Acid/toxicity , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Status Epilepticus/metabolism , Nerve Regeneration , Disease Models, Animal , Mice , Cell Movement
12.
Neurochem Res ; 49(7): 1782-1793, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38555337

ABSTRACT

Convulsive status epilepticus (CSE) is a common critical neurological condition that can lead to irreversible hippocampal neuron damage and cognitive dysfunction. Multiple studies have demonstrated the critical roles that long non-coding RNA Mir155hg plays in a variety of diseases. However, less is known about the function and mechanism of Mir155hg in CSE. Here we investigate and elucidate the mechanism underlying the contribution of Mir155hg to CSE-induced hippocampal neuron injury. By applying high-throughput sequencing, we examined the expression of differentially expressed genes in normal and CSE rats. Subsequent RT-qPCR enabled us to measure the level of Mir155hg in rat hippocampal tissue. Targeted knockdown of Mir155hg was achieved by the AAV9 virus. Additionally, we utilized HE and Tunel staining to evaluate neuronal injury. Immunofluorescence (IF), Golgi staining, and brain path clamping were also used to detect the synaptic plasticity of hippocampal neurons. Finally, through IF staining and Sholl analysis, we assessed the degree of microglial phagocytic function. It was found that the expression of Mir155hg was elevated in CSE rats. HE and Tunel staining results showed that Mir155hg knockdown suppressed the hippocampal neuron loss and apoptosis followed CSE. IF, Golgi staining and brain path clamp data found that Mir155hg knockdown enhanced neuronal synaptic plasticity. The results from IF staining and Sholl analysis showed that Mir155hg knockdown enhanced microglial phagocytosis. Our findings suggest that Mir155hg promotes CSE-induced hippocampal neuron injury by inhibiting microglial phagocytosis.


Subject(s)
Hippocampus , MicroRNAs , Microglia , Neurons , Phagocytosis , Rats, Sprague-Dawley , Status Epilepticus , Animals , Status Epilepticus/metabolism , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Hippocampus/metabolism , Hippocampus/pathology , Microglia/metabolism , Neurons/metabolism , Male , Phagocytosis/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , Rats , Apoptosis/physiology , Neuronal Plasticity/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
13.
Mol Biol Rep ; 51(1): 292, 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38332381

ABSTRACT

BACKGROUND: Neuroinflammation contributes to both epileptogenesis and the associated neurodegeneration, so regulation of inflammatory signaling is a potential strategy for suppressing epilepsy development and pathological progression. Exosomes are enriched in microRNAs (miRNAs), considered as vital communication tools between cells, which have been proven as potential therapeutic method for neurological disease. Here, we investigated the role of miR129-5p-loaded mesenchymal stem cell (MSC)-derived exosomes in status epilepticus (SE) mice model. METHODS: Mice were divided into four groups: untreated control (CON group), kainic acid (KA)-induced SE groups (KA group), control exosome injection (KA + Exo-con group), miR129-5p-loaded exosome injection (KA + Exo-miR129-5p group). Hippocampal expression levels of miR129-5p, HMGB1, and TLR4 were compared among groups. Nissl and Fluoro-jade B staining were conducted to evaluate neuronal damage. In addition, immunofluorescence staining for IBA-1 and GFAP was performed to assess glial cell activation, and inflammatory factor content was determined by ELISA. Hippocampal neurogenesis was assessed by BrdU staining. RESULTS: The expression of HMGB1 was increased after KA-induced SE and peaking at 48 h, while hippocampal miR129-5p expression decreased in SE mice. Exo-miR129-5p injection reversed KA-induced upregulation of hippocampal HMGB1 and TLR4, alleviated neuronal damage in the hippocampal CA3, reduced IBA-1 + and GFAP + staining intensity, suppressed SE-associated increases in inflammatory factors, and decreased BrdU + cell number in dentate gyrus. CONCLUSIONS: Exosomes loaded with miR129-5p can protect neurons against SE-mediated degeneration by inhibiting the pro-inflammatory HMGB1/TLR4 signaling axis.


Subject(s)
Exosomes , HMGB1 Protein , MicroRNAs , Status Epilepticus , Animals , Mice , Bromodeoxyuridine/adverse effects , Bromodeoxyuridine/metabolism , Exosomes/metabolism , Hippocampus/metabolism , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Kainic Acid/adverse effects , Kainic Acid/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Neuroinflammatory Diseases , Seizures/genetics , Status Epilepticus/chemically induced , Status Epilepticus/genetics , Status Epilepticus/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
14.
Eur J Pharmacol ; 966: 176375, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38307381

ABSTRACT

The role of the Wnt/ß-catenin signaling pathway in epilepsy and the effects of its modulators as efficacious treatment options, though postulated, has not been sufficiently investigated. We evaluated the involvement of ß-catenin and GSK-3ß, the significant proteins in this pathway, in the lithium chloride-pilocarpine-induced status epilepticus model in rodents to study acute phase of temporal lobe epilepsy (TLE). The modulators studied were 6-BIO, a GSK-3ß inhibitor and Sulindac, a Dvl protein inhibitor. The disease group exhibited increased seizure score and seizure frequency, and the assessment of neurobehavioral parameters indicated notable alterations. Furthermore, histopathological examination of hippocampal brain tissues revealed significant neurodegeneration. Immunohistochemical study of hippocampus revealed neurogenesis in 6-BIO and sulindac groups. The gene and protein expression by RT-qPCR and western blotting studies indicated Wnt/ß-catenin pathway downregulation and increased apoptosis in the acute phase of TLE. 6-BIO was very efficient in upregulating the Wnt pathway, decreasing neuronal damage, increasing neurogenesis in hippocampus and decreasing seizure score and frequency in comparison to sulindac. This suggests that both GSK-3ß and ß-catenin are potential and novel drug targets for acute phase of TLE, and treatment options targeting these proteins could be beneficial in successfully managing acute epilepsy. Further evaluation of 6-BIO to explore its therapeutic potential in other models of epilepsy should be conducted.


Subject(s)
Epilepsy, Temporal Lobe , Status Epilepticus , Rats , Animals , Pilocarpine , Wnt Signaling Pathway/physiology , Lithium/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , beta Catenin/metabolism , Sulindac/adverse effects , Sulindac/metabolism , Hippocampus/metabolism , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Status Epilepticus/metabolism , Seizures/chemically induced , Seizures/drug therapy , Seizures/metabolism , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/drug therapy
15.
Int J Mol Sci ; 25(3)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38338969

ABSTRACT

In humans and animal models, temporal lobe epilepsy (TLE) is associated with reorganization of hippocampal neuronal networks, gliosis, neuroinflammation, and loss of integrity of the blood-brain barrier (BBB). More than 30% of epilepsies remain intractable, and characterization of the molecular mechanisms involved in BBB dysfunction is essential to the identification of new therapeutic strategies. In this work, we induced status epilepticus in rats through injection of the proconvulsant drug pilocarpine, which leads to TLE. Using RT-qPCR, double immunohistochemistry, and confocal imaging, we studied the regulation of reactive glia and vascular markers at different time points of epileptogenesis (latent phase-3, 7, and 14 days; chronic phase-1 and 3 months). In the hippocampus, increased expression of mRNA encoding the glial proteins GFAP and Iba1 confirmed neuroinflammatory status. We report for the first time the concomitant induction of the specific proteins CD31, PDGFRß, and ColIV-which peak at the same time points as inflammation-in the endothelial cells, pericytes, and basement membrane of the BBB. The altered expression of these proteins occurs early in TLE, during the latent phase, suggesting that they could be associated with the early rupture and pathogenicity of the BBB that will contribute to the chronic phase of epilepsy.


Subject(s)
Blood-Brain Barrier , Epilepsy, Temporal Lobe , Epilepsy , Receptor, Platelet-Derived Growth Factor beta , Status Epilepticus , Animals , Humans , Rats , Blood-Brain Barrier/metabolism , Collagen/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Epilepsy/metabolism , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/metabolism , Hippocampus/metabolism , Neuroglia/metabolism , Pericytes/metabolism , Pilocarpine/adverse effects , Rats, Sprague-Dawley , Status Epilepticus/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism
16.
Glia ; 72(6): 1136-1149, 2024 06.
Article in English | MEDLINE | ID: mdl-38406970

ABSTRACT

Sirtuin3 (Sirt3) is a nicotinamide adenine dinucleotide enzyme that contributes to aging, cancer, and neurodegenerative diseases. Recent studies have reported that Sirt3 exerts anti-inflammatory effects in several neuropathophysiological disorders. As epilepsy is a common neurological disease, in the present study, we investigated the role of Sirt3 in astrocyte activation and inflammatory processes after epileptic seizures. We found the elevated expression of Sirt3 within reactive astrocytes as well as in the surrounding cells in the hippocampus of patients with temporal lobe epilepsy and a mouse model of pilocarpine-induced status epilepticus (SE). The upregulation of Sirt3 by treatment with adjudin, a potential Sirt3 activator, alleviated SE-induced astrocyte activation; whereas, Sirt3 deficiency exacerbated astrocyte activation in the hippocampus after SE. In addition, our results showed that Sirt3 upregulation attenuated the activation of Notch1 signaling, nuclear factor kappa B (NF-κB) activity, and the production of interleukin-1ß (IL1ß) in the hippocampus after SE. By contrast, Sirt3 deficiency enhanced the activity of Notch1/NF-κB signaling and the production of IL1ß. These findings suggest that Sirt3 regulates astrocyte activation by affecting the Notch1/NF-κB signaling pathway, which contributes to the inflammatory response after SE. Therefore, therapies targeting Sirt3 may be a worthy direction for limiting inflammatory responses following epileptic brain injury.


Subject(s)
Epilepsy , Sirtuin 3 , Status Epilepticus , Animals , Humans , Mice , Astrocytes/metabolism , Epilepsy/metabolism , Hippocampus/metabolism , NF-kappa B/metabolism , Signal Transduction , Sirtuin 3/metabolism , Status Epilepticus/chemically induced , Status Epilepticus/metabolism
17.
Talanta ; 268(Pt 1): 125302, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37826935

ABSTRACT

Epilepsy is a prevalent neurological disorder with a complex pathogenesis and unpredictable nature, presenting limited treatment options in >30 % of affected individuals. Neurometabolic abnormalities have been observed in epilepsy patients, suggesting a disruption in the coupling between neural activity and energy metabolism in the brain. In this study, we employed amperometric biosensors based on a modified carbon fiber microelectrode platform to directly and continuously measure lactate and oxygen dynamics in the brain extracellular space. These biosensors demonstrated high sensitivity, selectivity, and rapid response time, enabling in vivo measurements with high temporal and spatial resolution. In vivo recordings in the cortex of anaesthetized rats revealed rapid and multiphasic fluctuations in extracellular lactate and oxygen levels following neuronal stimulation with high potassium. Furthermore, real-time measurement of lactate and oxygen concentration dynamics concurrently with network electrical activity during status epilepticus induced by 4-aminopyridine (4-AP) demonstrated phasic changes in lactate levels that correlated with bursts of electrical activity, while tonic levels of lactate remained stable during seizures. This study highlights the complex interplay between lactate dynamics, electrical activity, and oxygen utilization in epileptic seizures.


Subject(s)
Biosensing Techniques , Epilepsy , Status Epilepticus , Humans , Rats , Animals , Lactic Acid/metabolism , Oxygen , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Brain/metabolism , Seizures/metabolism , 4-Aminopyridine
18.
Glia ; 72(2): 274-288, 2024 02.
Article in English | MEDLINE | ID: mdl-37746760

ABSTRACT

Auditory dysfunction and increased neuronal activity in the auditory pathways have been reported in patients with temporal lobe epilepsy, but the cellular mechanisms involved are unknown. Here, we report that microglia play a role in the disinhibition of auditory pathways after status epilepticus in mice. We found that neuronal activity in the auditory pathways, including the primary auditory cortex and the medial geniculate body (MGB), was increased and auditory discrimination was impaired after status epilepticus. We further demonstrated that microglia reduced inhibitory synapses on MGB relay neurons over an 8-week period after status epilepticus, resulting in auditory pathway hyperactivity. In addition, we found that local removal of microglia from the MGB attenuated the increase in c-Fos+ relay neurons and improved auditory discrimination. These findings reveal that thalamic microglia are involved in auditory dysfunction in epilepsy.


Subject(s)
Microglia , Status Epilepticus , Mice , Humans , Animals , Geniculate Bodies/metabolism , Thalamus , Auditory Pathways/metabolism , Status Epilepticus/metabolism
19.
Turk J Med Sci ; 53(5): 1058-1066, 2023.
Article in English | MEDLINE | ID: mdl-38813010

ABSTRACT

Background/aim: Propofol is a positive allosteric modulator of GABAA receptor (GABAAR) and has potent antioxidant activity. The aim of this study was to investigate the effect of propofol on damage to the cerebral cortex and hippocampus in a lithium chloride (LiCl)-pilocarpine animal model of status epilepticus (SE). Materials and methods: Adult male Sprague Dawley rats were injected with LiCl-pilocarpine to induce SE. They were then randomized and injected 30 min later with vehicle saline (SE+saline), propofol (SE+PPF, 50 mg/kg), Diazepam (SE+DZP, 10 mg/kg), Scopolamine (SE+SCOP, 10 mg/kg), or MK-801 (SE+MK-801, 2 mg/kg). Another group of rats received saline only and served as the naïve control (BLK). The levels of superoxide dismutase (SOD), glutathione (GSH) and malondialdehyde (MDA) in the serum, cortex and hippocampus were analyzed 2 and 24 h posttreatment. The degree of tissue damage in the cortex and hippocampus of individual rats was assessed 24 h posttreatment, together with expression of the GABAAR α1 subunit. Results: The propofol group showed reduced levels of tissue damage in the cerebral cortex and hippocampus, decreased levels of MDA, and increased levels of GSH compared to the SE+saline group. No changes in SOD level were observed in serum and tissue samples from the cortex and hippocampus of SE+saline rats. Immunohistochemistry and Western blot assays showed that propofol treatment significantly increased the expression of GABAAR α1 subunit in the cortical and hippocampal tissues of SE rats. Conclusion: Propofol treatment protected against SE-induced tissue injury in the cortex and hippocampus of rats. This was due at least in part to its antioxidant activity and to its induction of GABAAR α1 subunit expression in the brain.


Subject(s)
Disease Models, Animal , Lithium Chloride , Oxidative Stress , Pilocarpine , Propofol , Rats, Sprague-Dawley , Receptors, GABA-A , Status Epilepticus , Animals , Propofol/pharmacology , Receptors, GABA-A/metabolism , Receptors, GABA-A/drug effects , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Status Epilepticus/drug therapy , Pilocarpine/toxicity , Male , Lithium Chloride/pharmacology , Oxidative Stress/drug effects , Rats , Hippocampus/metabolism , Hippocampus/drug effects , Brain Injuries/metabolism , Brain Injuries/drug therapy , Brain Injuries/chemically induced , Malondialdehyde/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects
20.
Neurología (Barc., Ed. impr.) ; 33(9): 577-582, nov.-dic. 2018. graf
Article in Spanish | IBECS | ID: ibc-176003

ABSTRACT

INTRODUCCIÓN: El status epilepticus (SE) es un tipo de actividad epiléptica que causa atrofia cerebelar y pérdida de células de Purkinje en humanos y en animales de experimentación. El cerebelo es una región con alto contenido de ácido gama-aminobutírico (GABA) y glutamato, y algunos estudios refieren cambios en su concentración después de las convulsiones. Sin embargo, hasta la fecha no existen estudios que hayan analizado su efecto en diferentes regiones cerebelares en ratas en desarrollo. El objetivo del presente estudio fue realizar un curso temporal del efecto del SE inducido en ratas Wistar de 14 días de edad (P14) sobre el contenido tisular de GABA y glutamato en el vermis y los hemisferios cerebelares. MÉTODOS: El SE se indujo con el modelo de litio-pilocarpina; las ratas control se inyectaron con salina. Seis h, 24 h o 30 días después del inicio del SE o de la aplicación de solución salina, las ratas se anestesiaron y decapitaron, se extrajo su cerebelo y se separaron el vermis y los hemisferios. Las ratas de ambos grupos se anestesiaron y decapitaron, se extrajo su cerebelo y se separaron el vermis y los hemisferios. Ambas regiones se homogeneizaron (ácido perclórico 0,1 M conteniendo metabisulfito de sodio 4 mM) y centrifugaron, y el sobrenadante se empleó para cuantificar la concentración tisular de GABA y glutamato por cromatografía de líquidos de alta resolución acoplada a un detector fluorométrico. RESULTADOS: El SE no modificó la concentración de GABA y glutamato a los diferentes tiempos de análisis ni en el vermis ni en los hemisferios cerebelares. CONCLUSIONES: El cerebelo en desarrollo es resistente a los cambios neuroquímicos a corto y largo plazo producidos por el SE


INTRODUCTION: Status epilepticus (SE) is an epileptic condition that can cause cerebellar atrophy and loss of Purkinje cells in both humans and research animals. Cerebellum is a region rich in γ-aminobutyric acid (GABA) and glutamate, and some studies have shown that their concentrations may be altered after convulsions. However, there are no studies showing the effect of seizures on different cerebellar regions in developing rats. Time course of the effect of status epilepticus induced in the developing rat on γ-amino butyric acid and glutamate cerebellar concentration. METHODS: SE was induced using the lithium-pilocarpine model; control rats were injected with saline solution. At 6h, 24h, and 1 month after SE o saline injection, rats were anaesthetised with pentobarbital and decapitated, and cerebella were extracted. The vermis and hemispheres were dissected and homogenised in 0.1M perchloric acid containing 4mM sodium bisulfite. Homogenates were centrifuged and supernatant was used to quantify GABA, and glutamate tissue concentrations by HPLC coupled with fluorometric detection. RESULTS: SE did not alter GABA and glutamate tissue concentration in the cerebellar vermis and hemispheres. CONCLUSION: The developing rat cerebellum is resistant to both short- and long-term neurochemical changes induced by SE


Subject(s)
Animals , Male , Rats , Cerebellum/metabolism , Glutamic Acid/metabolism , Status Epilepticus/metabolism , gamma-Aminobutyric Acid/metabolism , Cerebellum , Growth and Development , Rats, Wistar , Status Epilepticus/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL